Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Methods Mol Biol ; 2769: 27-55, 2024.
Article in English | MEDLINE | ID: mdl-38315387

ABSTRACT

The hepatotoxic N-nitroso compound diethylnitrosamine (DEN) administered intraperitoneally (i.p.) induces liver neoplasms in rodents that reproducibly recapitulate some aspects of human hepatocarcinogenesis. In particular, DEN drives the stepwise formation of pre-neoplastic and neoplastic (benign or malignant) hepatocellular lesions reminiscent of the initiation-promotion-progression sequence typical of chemical carcinogenesis. In humans, the development of hepatocellular carcinoma (HCC) is also a multi-step process triggered by continuous hepatocellular injury, chronic inflammation, and compensatory hyperplasia that fuel the emergence of dysplastic liver lesions followed by the formation of early HCC. The DEN-induced liver tumorigenesis model represents a versatile preclinical tool that enables the study of many tumor development modifiers (genetic background, gene knockout or overexpression, diets, pollutants, or drugs) with a thorough follow-up of the multistage process on live animals by means of high-resolution imaging. Here, we provide a comprehensive protocol for the induction of hepatocellular neoplasms in wild-type C57BL/6J male mice following i.p. DEN injection (25 mg/kg) at 14 days of age and 36 weeks feeding of a high-fat high-sucrose (HFHS) diet. We emphasize the use of ultrasound liver imaging to follow tumor development and provide histopathological correlations. We also discuss the extrinsic and intrinsic factors known to modify the course of liver tumorigenesis in this model.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Male , Mice , Animals , Infant , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/diagnostic imaging , Diethylnitrosamine/toxicity , Mice, Inbred C57BL , Carcinogenesis/pathology , Diet, High-Fat/adverse effects , Liver/diagnostic imaging , Liver/pathology , Ultrasonography
2.
Cancers (Basel) ; 15(14)2023 Jul 22.
Article in English | MEDLINE | ID: mdl-37509384

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and one of the deadliest cancers worldwide. Despite extensive research, the biological mechanisms underlying HCC's development and progression remain only partially understood. Chronic overeating and/or sedentary-lifestyle-associated obesity, which promote Non-Alcoholic Fatty Liver Disease (NAFLD), have recently emerged as worrying risk factors for HCC. NAFLD is characterized by excessive hepatocellular lipid accumulation (steatosis) and affects one quarter of the world's population. Steatosis progresses in the more severe inflammatory form, Non-Alcoholic Steatohepatitis (NASH), potentially leading to HCC. The incidence of NASH is expected to increase by up to 56% over the next 10 years. Better diagnoses and the establishment of effective treatments for NAFLD and HCC will require improvements in our understanding of the fundamental mechanisms of the disease's development. This review describes the pathogenesis of NAFLD and the mechanisms underlying the transition from NAFL/NASH to HCC. We also discuss a selection of appropriate preclinical models of NAFLD for research, from cellular models such as liver-on-a-chip models to in vivo models, focusing particularly on mouse models of dietary NAFLD-HCC.

3.
Dev Cell ; 57(14): 1728-1741.e6, 2022 07 25.
Article in English | MEDLINE | ID: mdl-35768000

ABSTRACT

Non-alcoholic steatotic liver disease (NAFLD) is the most common cause of chronic liver disease worldwide. NAFLD has a major effect on the intrinsic proliferative properties of hepatocytes. Here, we investigated the mechanisms underlying the activation of DNA damage response during NAFLD. Proliferating mouse NAFLD hepatocytes harbor replication stress (RS) with an alteration of the replication fork's speed and activation of ATR pathway, which is sufficient to cause DNA breaks. Nucleotide pool imbalance occurring during NAFLD is the key driver of RS. Remarkably, DNA lesions drive cGAS/STING pathway activation, a major component of cells' intrinsic immune response. The translational significance of this study was reiterated by showing that lipid overload in proliferating HepaRG was sufficient to induce RS and nucleotide pool imbalance. Moreover, livers from NAFLD patients displayed nucleotide pathway deregulation and cGAS/STING gene alteration. Altogether, our findings shed light on the mechanisms by which damaged NAFLD hepatocytes might promote disease progression.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , DNA Damage , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Nucleotides , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism
4.
Cancers (Basel) ; 13(20)2021 Oct 14.
Article in English | MEDLINE | ID: mdl-34680300

ABSTRACT

Polyploidy, also known as whole-genome amplification, is a condition in which the organism has more than two basic sets of chromosomes. Polyploidy frequently arises during tissue development and repair, and in age-associated diseases, such as cancer. Its consequences are diverse and clearly different between systems. The liver is a particularly fascinating organ in that it can adapt its ploidy to the physiological and pathological context. Polyploid hepatocytes are characterized in terms of the number of nuclei per cell (cellular ploidy; mononucleate/binucleate hepatocytes) and the number of chromosome sets in each nucleus (nuclear ploidy; diploid, tetraploid, octoploid). The advantages and disadvantages of polyploidy in mammals are not fully understood. About 30% of the hepatocytes in the human liver are polyploid. In this review, we explore the mechanisms underlying the development of polyploid cells, our current understanding of the regulation of polyploidization during development and pathophysiology and its consequences for liver function. We will also provide data shedding light on the ways in which polyploid hepatocytes cope with centrosome amplification. Finally, we discuss recent discoveries highlighting the possible roles of liver polyploidy in protecting against tumor formation, or, conversely, contributing to liver tumorigenesis.

5.
J Hepatol ; 74(6): 1386-1397, 2021 06.
Article in English | MEDLINE | ID: mdl-33484773

ABSTRACT

BACKGROUND & AIMS: The NKG2D system is a potent immunosurveillance mechanism in cancer, wherein the activating NK cell receptor (NKG2D) on immune cells recognises its cognate ligands on tumour cells. Herein, we evaluated the expression of NKG2D ligands in hepatocellular carcinoma (HCC), in both humans and mice, taking the genomic features of HCC tumours into account. METHODS: The expression of NKG2D ligands (MICA, MICB, ULBP1 and ULBP2) was analysed in large human HCC datasets by Fluidigm TaqMan and RNA-seq methods, and in 2 mouse models (mRNA and protein levels) reproducing the features of both major groups of human tumours. RESULTS: We provide compelling evidence that expression of the MICA and MICB ligands in human HCC is associated with tumour aggressiveness and poor patient outcome. We also found that the expression of ULBP1 and ULBP2 was associated with poor patient outcome, and was downregulated in CTNNB1-mutated HCCs displaying low levels of inflammation and associated with a better prognosis. We also found an inverse correlation between ULBP1/2 expression levels and the expression of ß-catenin target genes in patients with HCC, suggesting a role for ß-catenin signalling in inhibiting expression. We showed in HCC mouse models that ß-catenin signalling downregulated the expression of Rae-1 NKG2D ligands, orthologs of ULBPs, through TCF4 binding. CONCLUSIONS: We demonstrate that the expression of NKG2D ligands is associated with aggressive liver tumorigenesis and that the downregulation of these ligands by ß-catenin signalling may account for the less aggressive phenotype of CTNNB1-mutated HCC tumours. LAY SUMMARY: The NKG2D system is a potent immunosurveillance mechanism in cancer. However, its role in hepatocellular carcinoma development has not been widely investigated. Herein, we should that the expression of NKG2D ligands by tumour cells is associated with a more aggressive tumour subtype.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class I/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/metabolism , Signal Transduction/genetics , beta Catenin/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cohort Studies , Disease Models, Animal , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Histocompatibility Antigens Class I/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Prognosis , Young Adult
6.
Nat Rev Gastroenterol Hepatol ; 17(7): 391-405, 2020 07.
Article in English | MEDLINE | ID: mdl-32242122

ABSTRACT

Polyploidy (or whole-genome duplication) is the condition of having more than two basic sets of chromosomes. Polyploidization is well tolerated in many species and can lead to specific biological functions. In mammals, programmed polyploidization takes place during development in certain tissues, such as the heart and placenta, and is considered a feature of differentiation. However, unscheduled polyploidization can cause genomic instability and has been observed in pathological conditions, such as cancer. Polyploidy of the liver parenchyma was first described more than 100 years ago. The liver is one of the few mammalian organs that display changes in polyploidy during homeostasis, regeneration and in response to damage. In the human liver, approximately 30% of hepatocytes are polyploid. The polyploidy of hepatocytes results from both nuclear polyploidy (an increase in the amount of DNA per nucleus) and cellular polyploidy (an increase in the number of nuclei per cell). In this Review, we discuss the regulation of polyploidy in liver development and pathophysiology. We also provide an overview of current knowledge about the mechanisms of hepatocyte polyploidization, its biological importance and the fate of polyploid hepatocytes during liver tumorigenesis.


Subject(s)
Hepatocytes/physiology , Liver/embryology , Liver/physiopathology , Polyploidy , Animals , Cell Differentiation , Homeostasis , Humans , Liver/pathology
7.
Gut ; 69(2): 355-364, 2020 02.
Article in English | MEDLINE | ID: mdl-30979717

ABSTRACT

OBJECTIVES: Polyploidy is a fascinating characteristic of liver parenchyma. Hepatocyte polyploidy depends on the DNA content of each nucleus (nuclear ploidy) and the number of nuclei per cell (cellular ploidy). Which role can be assigned to polyploidy during human hepatocellular carcinoma (HCC) development is still an open question. Here, we investigated whether a specific ploidy spectrum is associated with clinical and molecular features of HCC. DESIGN: Ploidy spectra were determined on surgically resected tissues from patients with HCC as well as healthy control tissues. To define ploidy profiles, a quantitative and qualitative in situ imaging approach was used on paraffin tissue liver sections. RESULTS: We first demonstrated that polyploid hepatocytes are the major components of human liver parenchyma, polyploidy being mainly cellular (binuclear hepatocytes). Across liver lobules, polyploid hepatocytes do not exhibit a specific zonation pattern. During liver tumorigenesis, cellular ploidy is drastically reduced; binuclear polyploid hepatocytes are barely present in HCC tumours. Remarkably, nuclear ploidy is specifically amplified in HCC tumours. In fact, nuclear ploidy is amplified in HCCs harbouring a low degree of differentiation and TP53 mutations. Finally, our results demonstrated that highly polyploid tumours are associated with a poor prognosis. CONCLUSIONS: Our results underline the importance of quantification of cellular and nuclear ploidy spectra during HCC tumorigenesis.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Polyploidy , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/surgery , Case-Control Studies , Cell Differentiation/genetics , Cell Nucleus/pathology , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Female , Hepatocytes/pathology , Humans , Liver Neoplasms/pathology , Liver Neoplasms/surgery , Male , Middle Aged , Prognosis , Young Adult
8.
Sci Rep ; 9(1): 14614, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31601995

ABSTRACT

Mammalian p38α MAPK (Mitogen-Activated Protein Kinase) transduces a variety of extracellular signals that regulate cellular processes, such as inflammation, differentiation, proliferation or apoptosis. In the liver, depending of the physiopathological context, p38α acts as a negative regulator of hepatocyte proliferation as well as a promotor of inflammatory processes. However, its function during an acute injury, in adult liver, remains uncharacterized. In this study, using mice that are deficient in p38α specifically in mature hepatocytes, we unexpectedly found that lack of p38α protected against acute injury induced by CCl4 compound. We demonstrated that the hepatoprotective effect alleviated ROS accumulation and shaped the inflammatory response to promote efficient tissue repair. Mechanistically, we provided strong evidence that Ccl2/Ccl5 chemokines were crucial for a proper hepatoprotective response observed secondary to p38α ablation. Indeed, antibody blockade of Ccl2/Ccl5 was sufficient to abrogate hepatoprotection through a concomitant decrease of both inflammatory cells recruitment and antioxidative response that result ultimately in higher liver damages. Our findings suggest that targeting p38α expression and consequently orientating immune response may represent an attractive approach to favor tissue recovery after acute liver injury.


Subject(s)
Liver Regeneration , Liver/drug effects , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/metabolism , Animals , Antioxidants/metabolism , Apoptosis , Carbon Tetrachloride/adverse effects , Cell Differentiation , Cell Proliferation , Chemokine CCL2/metabolism , Chemokine CCL5/metabolism , Crosses, Genetic , Female , Gene Deletion , Gene Expression Profiling , Hepatocytes , Inflammation , Liver/injuries , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reactive Oxygen Species/metabolism
9.
Med Sci (Paris) ; 35(6-7): 519-526, 2019.
Article in French | MEDLINE | ID: mdl-31274081

ABSTRACT

Polyploidy (alias whole genome amplification) refers to organisms containing more than two basic sets of chromosomes. Polyploidy was first observed in plants more than a century ago, and it is known that such processes occur in many eukaryotes under a variety of circumstances. In mammals, the development of polyploid cells can contribute to tissue differentiation and therefore possibly a gain of function. Alternately, it can be associated with development of disease such as cancer. Polyploidy can occur because of cell fusion or abnormal cell division. Polyploidy is a common characteristic of the mammalian liver. Polyploidization occurs notably during liver development, but also in adults because of cellular stress. Recent progresses have unraveled the mechanisms and functional consequences of hepatocytes polyploidization during normal and pathological liver growth.


Subject(s)
Liver/metabolism , Liver/pathology , Polyploidy , Adult , Animals , Cell Division/genetics , Cell Fusion , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver/cytology
10.
Hepatology ; 69(1): 160-178, 2019 01.
Article in English | MEDLINE | ID: mdl-30070727

ABSTRACT

Leukocyte cell-derived chemotaxin-2 (LECT2) was originally identified as a hepatocyte-secreted chemokine-like factor and a positive target of ß-catenin signaling. Here, we dissected out the mechanisms by which LECT2 modulates hepatocellular carcinoma (HCC) development using both HCC mouse models and human HCC samples. We have demonstrated that LECT2 exhibits dual abilities as it has profound repercussions on the tumor phenotype itself and the immune microenvironment. Its absence confers Ctnnb-1-mutated tumor hepatocytes a stronger ability to undergo epithelial to mesenchymal transition and fosters the accumulation of pejorative inflammatory monocytes harboring immunosuppressive properties and strong tumor-promoting potential. Consistent with our HCC mouse model, a low level of LECT2 in human HCC is strongly associated with high tumor grade and the presence of inflammatory infiltrates, emphasizing the clinical value of LECT2 in human liver tumorigenesis. Conclusion: Our findings have demonstrated that LECT2 is a key player in liver tumorigenesis because its absence reshapes the tumor microenvironment and the tumor phenotype, revealing LECT2 as a promising immunotherapeutic option for HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Monocytes/physiology , Receptors, G-Protein-Coupled/physiology , Receptors, Peptide/physiology , Animals , Carcinoma, Hepatocellular/etiology , Disease Models, Animal , Disease Progression , Humans , Inflammation/complications , Liver Neoplasms/etiology , Mice , Tumor Cells, Cultured
11.
Cell Rep ; 22(8): 1994-2005, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29466728

ABSTRACT

Liver kinase B1 (LKB1) is involved in several biological processes and is a key regulator of hepatic metabolism and polarity. Here, we demonstrate that the master kinase LKB1 plays a dual role in liver regeneration, independently of its major target, AMP-activated protein kinase (AMPK). We found that the loss of hepatic Lkb1 expression promoted hepatocyte proliferation acceleration independently of metabolic/energetic balance. LKB1 regulates G0/G1 progression, specifically by controlling epidermal growth factor receptor (EGFR) signaling. Furthermore, later in regeneration, LKB1 controls mitotic fidelity. The deletion of Lkb1 results in major alterations to mitotic spindle formation along the polarity axis. Thus, LKB1 deficiency alters ploidy profile at late stages of regeneration. Our findings highlight the dual role of LKB1 in liver regeneration, as a guardian of hepatocyte proliferation and genomic integrity.


Subject(s)
Genome , Hepatocytes/cytology , Hepatocytes/metabolism , Liver Regeneration/genetics , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Animals , Cell Proliferation , Enzyme Activation , ErbB Receptors/metabolism , Gene Deletion , Gene Silencing , Liver/cytology , Mice , Mitosis , Ploidies , Protein Serine-Threonine Kinases/deficiency , Signal Transduction
12.
Clin Res Hepatol Gastroenterol ; 41(2): 123-125, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28139382

ABSTRACT

Polyploidy is defined as an increase in genome DNA content and is observed in all mammalian species. Polyploidy is a common characteristic of hepatocytes. Polyploidization occurs mainly during liver development, but also in adults with increasing age or due to cellular stress. During liver development, hepatocytes polyploidization occurs through cytokinesis failure leading to the genesis of binucleate hepatocytes. Recently, Hsu et al. demonstrated that miR-122 is a key regulator of hepatic binucleation. In fact, during liver development, miR-122 directly antagonizes procytokinesis targets and thus induces cytokinesis failure leading to the genesis of binucleate hepatocytes.


Subject(s)
Hepatocytes/ultrastructure , Liver/ultrastructure , MicroRNAs/physiology , Polyploidy , Animals , Cell Nucleus/physiology , Humans , Liver/growth & development
13.
J Clin Invest ; 125(3): 981-92, 2015 Mar 02.
Article in English | MEDLINE | ID: mdl-25621497

ABSTRACT

Polyploidization is one of the most dramatic changes that can occur in the genome. In the liver, physiological polyploidization events occur during both liver development and throughout adult life. Here, we determined that a pathological polyploidization takes place in nonalcoholic fatty liver disease (NAFLD), a widespread hepatic metabolic disorder that is believed to be a risk factor for hepatocellular carcinoma (HCC). In murine models of NAFLD, the parenchyma of fatty livers displayed alterations of the polyploidization process, including the presence of a large proportion of highly polyploid mononuclear cells, which are rarely observed in normal hepatic parenchyma. Biopsies from patients with nonalcoholic steatohepatitis (NASH) revealed the presence of alterations in hepatocyte ploidy compared with tissue from control individuals. Hepatocytes from NAFLD mice revealed that progression through the S/G2 phases of the cell cycle was inefficient. This alteration was associated with activation of a G2/M DNA damage checkpoint, which prevented activation of the cyclin B1/CDK1 complex. Furthermore, we determined that oxidative stress promotes the appearance of highly polyploid cells, and antioxidant-treated NAFLD hepatocytes resumed normal cell division and returned to a physiological state of polyploidy. Collectively, these findings indicate that oxidative stress promotes pathological polyploidization and suggest that this is an early event in NAFLD that may contribute to HCC development.


Subject(s)
Liver/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Oxidative Stress , Polyploidy , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , DNA Damage , Diet, High-Fat/adverse effects , Hepatocytes/pathology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice, Inbred C57BL , Middle Aged , Non-alcoholic Fatty Liver Disease/pathology , Risk Factors
14.
J Hepatol ; 60(1): 152-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24012615

ABSTRACT

BACKGROUND: AMP-activated protein kinase (AMPK) is an evolutionarily conserved sensor of cellular energy status that contributes to restoration of energy homeostasis by slowing down ATP-consuming pathways and activating ATP-producing pathways. Unexpectedly, in different systems, AMPK is also required for proper cell division. In the current study, we evaluated the potential effect of the AMPK catalytic subunit, AMPKα1, on hepatocyte proliferation. METHODS: Hepatocyte proliferation was determined in AMPKα1 knockout and wild-type mice in vivo after two thirds partial hepatectomy, and in vitro in primary hepatocyte cultures. The activities of metabolic and cell cycle-related signaling pathways were measured. RESULTS: After partial hepatectomy, hepatocytes proliferated rapidly, correlating with increased AMPK phosphorylation. Deletion of AMPKα1 delayed liver regeneration by impacting on G1/S transition phase. The proliferative defect of AMPKα1-deficient hepatocytes was cell autonomous, and independent of energy balance. The priming phase, lipid droplet accumulation, protein anabolic responses and growth factor activation after partial hepatectomy occurred normally in the absence of AMPKα1 activity. By contrast, mRNA and protein expression of cyclin A2, a key driver of S phase progression, were compromised in the absence of AMPK activity. Importantly, AMPKα1 controlled cyclin A2 transcription mainly through the ATF/CREB element. CONCLUSIONS: Our study highlights a novel role for AMPKα1 as a positive regulator of hepatocyte division occurring independently of energy balance.


Subject(s)
AMP-Activated Protein Kinases/physiology , Cell Proliferation , Cyclin A2/physiology , Hepatocytes/physiology , Animals , Cyclin A2/genetics , Energy Metabolism , Liver Regeneration , Mice , Mice, Inbred C57BL , S Phase
15.
Int J Hepatol ; 2012: 282430, 2012.
Article in English | MEDLINE | ID: mdl-23150829

ABSTRACT

Most cells in mammalian tissues usually contain a diploid complement of chromosomes. However, numerous studies have demonstrated a major role of "diploid-polyploid conversion" during physiopathological processes in several tissues. In the liver parenchyma, progressive polyploidization of hepatocytes takes place during postnatal growth. Indeed, at the suckling-weaning transition, cytokinesis failure events induce the genesis of binucleated tetraploid liver cells. Insulin signalling, through regulation of the PI3K/Akt signalling pathway, is essential in the establishment of liver tetraploidization by controlling cytoskeletal organisation and consequently mitosis progression. Liver cell polyploidy is generally considered to indicate terminal differentiation and senescence, and both lead to a progressive loss of cell pluripotency associated to a markedly decreased replication capacity. Although adult liver is a quiescent organ, it retains a capacity to proliferate and to modulate its ploidy in response to various stimuli or aggression (partial hepatectomy, metabolic overload (i.e., high copper and iron hepatic levels), oxidative stress, toxic insult, and chronic hepatitis etc.). Here we review the mechanisms and functional consequences of hepatocytes polyploidization during normal and pathological liver growth.

16.
J Clin Invest ; 121(7): 2821-32, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21633171

ABSTRACT

Rapamycin is an antibiotic inhibiting eukaryotic cell growth and proliferation by acting on target of rapamycin (TOR) kinase. Mammalian TOR (mTOR) is thought to work through 2 independent complexes to regulate cell size and cell replication, and these 2 complexes show differential sensitivity to rapamycin. Here we combine functional genetics and pharmacological treatments to analyze rapamycin-sensitive mTOR substrates that are involved in cell proliferation and tissue regeneration after partial hepatectomy in mice. After hepatectomy, hepatocytes proliferated rapidly, correlating with increased S6 kinase phosphorylation, while treatment with rapamycin derivatives impaired regeneration and blocked S6 kinase activation. In addition, genetic deletion of S6 kinase 1 (S6K1) caused a delay in S phase entry in hepatocytes after hepatectomy. The proliferative defect of S6K1-deficient hepatocytes was cell autonomous, as it was also observed in primary cultures and hepatic overexpression of S6K1-rescued proliferation. We found that S6K1 controlled steady-state levels of cyclin D1 (Ccnd1) mRNA in liver, and cyclin D1 expression was required to promote hepatocyte cell cycle. Notably, in vivo overexpression of cyclin D1 was sufficient to restore the proliferative capacity of S6K-null livers. The identification of an S6K1-dependent mechanism participating in cell proliferation in vivo may be relevant for cancer cells displaying high mTOR complex 1 activity and cyclin D1 accumulation.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cell Proliferation/drug effects , Liver Regeneration/physiology , Liver/drug effects , Liver/physiology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Sirolimus/pharmacology , Animals , Cell Cycle/drug effects , Cell Cycle/physiology , Cells, Cultured , Cyclin D1/genetics , Cyclin D1/metabolism , Genotype , Hepatectomy , Hepatocytes/cytology , Hepatocytes/physiology , Isoenzymes/genetics , Isoenzymes/metabolism , Liver/cytology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes , Proteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics , TOR Serine-Threonine Kinases
17.
Curr Biol ; 21(6): 473-9, 2011 Mar 22.
Article in English | MEDLINE | ID: mdl-21376600

ABSTRACT

In eukaryotes, cytokinesis generally involves an actomyosin ring, the contraction of which promotes daughter cell segregation. Assembly of the contractile ring is tightly controlled in space and time. In the fission yeast, contractile ring components are first organized by the anillin-like protein Mid1 into medial cortical nodes. These nodes then coalesce laterally into a functional contractile ring. Although Mid1 is present at the medial cortex throughout G2, recruitment of contractile ring components to nodes starts only at mitotic onset, indicating that this event is cell-cycle regulated. Polo kinases are key temporal coordinators of mitosis and cytokinesis, and the Polo-like kinase Plo1 is known to activate Mid1 nuclear export at mitotic onset, coupling division plane specification to nuclear position. Here we provide evidence that Plo1 also triggers the recruitment of contractile ring components into medial cortical nodes. Plo1 binds at least two independent sites on Mid1, including a consensus site phosphorylated by Cdc2. Plo1 phosphorylates several residues within the first 100 amino acids of Mid1, which directly interact with the IQGAP Rng2, and influences the timing of myosin II recruitment. Plo1 thereby facilitates contractile ring assembly at mitotic onset.


Subject(s)
Actomyosin/physiology , Contractile Proteins/metabolism , Cytokinesis/physiology , Myosin Type II/metabolism , Protein Serine-Threonine Kinases/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/physiology , Actomyosin/metabolism , Binding Sites/genetics , CDC2 Protein Kinase/metabolism , Immunoprecipitation , Mass Spectrometry , Microscopy, Fluorescence , Phosphorylation , Plasmids/genetics , Schizosaccharomyces pombe Proteins/genetics , Time-Lapse Imaging
18.
Adv Exp Med Biol ; 676: 123-35, 2010.
Article in English | MEDLINE | ID: mdl-20687473

ABSTRACT

Eukaryotic organisms usually contain a diploid complement of chromosomes. However, there are a number of exceptions. Organisms containing an increase in DNA content by whole number multiples of the entire set of chromosomes are defined as polyploid. Cells that contain more than two sets of chromosomes were first observed in plants about a century ago and it is now recognized that polyploidy cells form in many eukaryotes under a wide variety of circumstance. Although it is less common in mammals, some tissues, including the liver, show a high percentage of polyploid cells. Thus, during postnatal growth, the liver parenchyma undergoes dramatic changes characterized by gradual polyploidization during which hepatocytes of several ploidy classes emerge as a result of modified cell-division cycles. This process generates the successive appearance of tetraploid and octoploid cell classes with one or two nuclei (mononucleated or binucleated). Liver cells polyploidy is generally considered to indicate terminal differentiation and senescence and to lead both to the progressive loss of cell pluripotency and a markedly decreased replication capacity. In adults, liver polyploidization is differentially regulated upon loss of liver mass and liver damage. Interestingly, partial hepatectomy induces marked cell proliferation followed by an increase in liver ploidy. In contrast, during hepatocarcinoma (HCC), growth shifts to a nonpolyploidizing pattern and expansion of the diploid hepatocytes population is observed in neoplastic nodules. Here we review the current state of understanding about how polyploidization is regulated during normal and pathological liver growth and detail by which mechanisms hepatocytes become polyploid.


Subject(s)
Chromosomes, Human/metabolism , Hepatocytes/metabolism , Liver Neoplasms/metabolism , Liver/metabolism , Polyploidy , Adult , Animals , Cell Differentiation , Cell Division , Cellular Senescence , Chromosomes, Human/genetics , Hepatectomy , Hepatocytes/pathology , Humans , Liver/pathology , Liver Neoplasms/pathology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Regeneration
19.
Cell Cycle ; 9(3): 460-6, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20090410

ABSTRACT

The formation of polyploid cells is part of the developmental program in several tissues. Polyploidy is a characteristic feature of mammalian hepatocytes and it is emerging that this process is an important mechanism of restricting liver growth. We previously demonstrated that during post-natal development, binucleated tetraploid hepatocytes arise due to a failure in cytokinesis. The genesis of such binucleated tetraploid cells is the crucial step for the establishment of liver polyploidization. Our recent work identified the cellular signaling pathway controlling this process. Rats with low levels of circulating insulin exhibit reduced formation of binucleated tetraploid hepatocytes, whereas rats injected with insulin exhibit increased formation of binucleated tetraploid hepatocytes. Furthermore, modulation of Akt activity clearly controls cytokinesis failure events indicating that the PI3K-Akt pathway, downstream from the insulin signal, is central to tetraploidization process. Here, we discuss these findings in the context of how cells become polyploid during physiological or pathological growth.


Subject(s)
Hepatocytes/cytology , Insulin/metabolism , Liver/cytology , Polyploidy , Signal Transduction , Animals , Animals, Newborn , Cytokinesis , Hepatocytes/metabolism , Humans , Liver/growth & development , Mice , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...